EP2265643B1 - Schema posologique pour traiter le psoriasis et l'arhtrite rhumatoide - Google Patents

Schema posologique pour traiter le psoriasis et l'arhtrite rhumatoide Download PDF

Info

Publication number
EP2265643B1
EP2265643B1 EP09727585.3A EP09727585A EP2265643B1 EP 2265643 B1 EP2265643 B1 EP 2265643B1 EP 09727585 A EP09727585 A EP 09727585A EP 2265643 B1 EP2265643 B1 EP 2265643B1
Authority
EP
European Patent Office
Prior art keywords
antibody
domain
dose
chain
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Not-in-force
Application number
EP09727585.3A
Other languages
German (de)
English (en)
Other versions
EP2265643A1 (fr
Inventor
Frank Osterroth
Silke Aigner
Matthias Germer
Christoph Uherek
Elmar Kraus
Andrea Wartenberg-Demand
Daniele Wolf
Sibylle Kaiser
Juergen Lindner
Christoph Bruecher
Benjamin Daelken
Gregor Schulz
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biotest AG
Original Assignee
Biotest AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GB0804686A external-priority patent/GB0804686D0/en
Priority claimed from GB0817811A external-priority patent/GB0817811D0/en
Application filed by Biotest AG filed Critical Biotest AG
Publication of EP2265643A1 publication Critical patent/EP2265643A1/fr
Application granted granted Critical
Publication of EP2265643B1 publication Critical patent/EP2265643B1/fr
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2812Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention is concerned with treatment of autoimmune diseases.
  • the invention involves a highly effective agent which is a humanised monoclonal antibody that may be administered to patients in lower dosages than previously known. It is particularly effective for patients having diseases or characteristics requiring lower doses for effective treatment.
  • the description envisages a pharmaceutical composition comprising the antibody in efficacious concentration, as well as uses and methods of treatment employing the compositions and medicaments comprising the antibody.
  • Autoimmunity is the failure of an organism to recognise its own constituent parts (down to sub-molecular levels) as "self", which results in an immune response against its own cells and tissues. Any disease that results from such an aberrant immune response is termed an autoimmune disease.
  • Autoimmune diseases include multiple sclerosis (MS), rheumatoid arthritis (RA), psoriasis, psoriatic arthritis, colitis ulcerosa, Crohn's disease, myasthenia gravis (MG), autoimmune polyglandular syndrome type II (APS-II), Hashimoto's thyroiditis (HT), type-1 diabetes (T1D), systemic lupus erythematosus (SLE) and autoimmune lymphoproliferative syndrome (ALS).
  • MS multiple sclerosis
  • RA rheumatoid arthritis
  • psoriasis psoriatic arthritis
  • colitis ulcerosa Crohn's disease
  • MG myasthenia gravis
  • Activation of 'autoreactive' T cells by presentation of autoantigens processed by antigen presenting cells (APC) leads to their clonal expansion and migration to the specific tissues, where they induce inflammation and tissue destruction.
  • APC antigen presenting cells
  • T cells are tolerant with regard to autologous tissue and only react on presentation of heterologous structures.
  • Central tolerance and peripheral tolerance comprise the two mechanisms by which the immune system hinders the autoreactive T cells from inducing their deleterious functions.
  • Central tolerance is mediated through negative selection. This process entails the elimination, through clonal deletion of autoreactive T cells, during ontogenic development in the thymus.
  • Peripheral tolerance is the backup available if central tolerance fails and autoreactive cells escape the thymus. This mechanism of tolerance occurs continuously throughout life, keeping autoreactive cells in check through immune ignorance (anergy), peripheral deletion and active suppression.
  • T regulatory cells (Tregs, formerly also designated “suppressor cells”) as part of active suppression maintain peripheral tolerance and regulate autoimmunity ( Suri-Payer et al., J Immunol. 157: 1799-1805 (1996 ); Asano et al., J Exp. Med. 184:387-396 (1996 ); Bonomo et al., J. Immunol. 154: 6602-6611 (1995 ); Willerford et al., Immunity 3: 521-530 (1995 ); Takahashi et al., Int. Immunol.
  • Tregs Several subsets of regulatory T cells have been characterized.
  • the family of Tregs consists of two key subsets: naturally arising e.g. CD4 + CD25 + Tregs and peripherally induced, Tr1 and Th3 Tregs.
  • NKTregs and CD8 + Tregs have been described in humans and rodents ( Fehérvari et al., J. Clin. Investigation 114: 1209-1217 (2004 )).
  • Thymus-derived Treg cells are the main regulatory cells involved, utilizing an array of TCRs targeted towards autoantigen recognition in order to maintain immune homeostasis in the periphery, and regulate autoimmunity and pathogenic immune responses.
  • Tregs The role of Tregs is exemplified best by experiments involving reconstitution of immunodeficient nude mice with CD4 + cells that were depleted of CD25 + cells.
  • CD4 + CD25 - reconstituted nude mice develop various organ-specific autoimmune diseases, such as gastritis, oophoritis, orchitis, and thyroiditis ( Suri-Payeret al.; J. Immunol. 160: 1212-1218 (1998 )).
  • CD4 + CD25 + subset prevents the onset of these diseases ( Sakaguchi et al., J Immunol. 155: 1151-1164 (1995 )).
  • the protective value of CD4 + CD25 + cells against organ-specific autoimmunity has also been shown in several other models of autoimmunity (e.g. autoimmune gastritis, prostatitis, oophoritis, glomerulonephritis, epidimytis and thyroiditis) caused by neonatal thymectomy performed 3 days after birth (d3Tx) or inflammatory bowel disease caused by reconstitution of SCID mice with CD45RBhigh, CD4 + CD25 - T cells.
  • Administration of anti-CD25 antibody in vivo in mice also induces organ-localised autoimmune disease.
  • CD4 + CD25 + Tregs inhibit polyclonal and antigen-specific T cell activation.
  • the suppression is mediated by a cell contact-dependent mechanism that requires activation of CD4 + CD25 + Tregs via the TCR but Tregs do not show a proliferative response upon TCR activation or stimulation with mitogenic antibodies (anergic) ( Shevach, Nature Rev. Immunol 2 : 389 (2002 ).
  • mitogenic antibodies anergic
  • Type 1 T regulatory (Tr1) cells and Th3 cells.
  • Tr1 Type 1 T regulatory
  • Tr1 cells were induced by repetitive stimulation of TCR in the presence of IL-10 and were shown to mainly down-regulate immune responses via the production of high levels of IL-10 and moderate amounts of TGF- ⁇ ( Chen et al., J. Immunol. 171: 733-744 (2003 )).
  • Th3 cells (identified in a model of EAE after oral delivery of antigen) produce high amounts of TGF- ⁇ and variable amounts of IL-4 and IL-10, and IL-4 was shown to be a key factor for the differentiation of Th3 cells, in contrast to Tr1 cells ( Chen et al., Science 265:1237-1240 (1994 )).
  • immunosuppressive drugs Suppression of T cell function by using immunosuppressive drugs is a principal therapeutic strategy that has been used successfully to treat autoimmune diseases.
  • these drugs induce a general immunosuppression due to their poor selectivity, resulting in inhibition of not only the harmful functions of the immune system, but also useful ones.
  • several risks like infection, cancer and drug toxicity may occur.
  • T cell function agents interfering with T cell function are therapeutic mainstays for various autoimmune diseases.
  • TGN 1412 Treatment with a total dose of 5-10 mg of TGN 1412 (0.1 mg anti-CD28 per kilogram of body weight) lead to a systemic inflammatory response with multiorgan failure within 90 minutes after receiving a single intravenous dose of the TGN 1412 ( Suntharalingam et al, N Engl. J Med. 2006 Sep 7;355(10):1018-28 ).
  • CD4 + T cells play a major part in initiating and maintaining autoimmunity. Accordingly, it has been proposed to use mAbs against CD4 + T cells surface molecules, and in particular anti-CD4 mAbs, as immunosuppressive agents. Although numerous clinical studies confirmed the potential interest of this approach, they also raised several issues to be addressed in order to make anti-CD4 mAbs more suitable for use in routine clinical practice.
  • CD4 mAbs Several different mechanisms of action for CD4 mAbs have been proposed including: (1) antagonism of CD4-MHC II interactions resulting in inhibition of T cell activation, (2) CD4 receptor modulation as determined by a decrease in cell surface expression of CD4, (3) partial signaling through the CD4 receptor in the absence of T cell receptor cross-linking which can suppress subsequent T cell activation and trigger CD4 T cell apoptotic death, (4) Fc-mediated complement-dependent cytotoxicity (CDC) or antibody-dependent cellular cytotoxicity (ADCC) leading to CD4 T cell depletion, and (5) stimulation of regulatory T cells.
  • CDC complement-dependent cytotoxicity
  • ADCC antibody-dependent cellular cytotoxicity
  • the B-F5 antibody (murine IgG1 anti-human CD4) was tested in different autoimmune diseases.
  • Rhadot et al. discloses the immunological follow-up of 17 patients with rheumatoid arthritis treated in vivo with an anti-T CD4+ monoclonal antibody (B-F5), (Clin. Exp. Rheumatol., 1992, 10(4):365-374 ).
  • Wendling et al. discloses the treatment of rheumatoid arthritis with anti CD4 monoclonal antibody; open study of 25 patients with the B-F5 clone (Clin. Rheumatol., 1992; 11(4):542-547 ).
  • MS multiple sclerosis
  • humanized antibodies obtained by grafting the complementarity-determining regions (CDRs) of a mouse monoclonal antibody, which determine the antigen-binding specificity, onto the framework regions (FRs) of a human immunoglobulin molecule.
  • the aim of humanization is to obtain a recombinant antibody having the same antigen-binding properties as the mouse monoclonal antibody from which the CDR sequences were derived, and far less immunogenic in humans.
  • substituting CDRs from the mouse antibody for the human CDRs in human frameworks is sufficient to transfer the antigen-binding properties (including not only the specificity, but also the affinity for antigen).
  • some FR residues are important for antigen binding, because they directly contact the antigen in the antibody-antigen complex, or because they influence the conformation of CDRs and thus their antigen binding performance.
  • hB-F5 humanized B-F5
  • compositions for parenteral administration of from 0.1-10 mg, preferably from 1-5 mg.
  • Dosage regimes envisaged are an intravenous 1 mg per day dose and a 5 mg every second day dose for rheumatoid arthritis patients over a period of 10 days.
  • Wijdenes et al. in an abstract and poster presented at the EULAR conference, June 2005. They described the treatment of 11 patients suffering from rheumatoid arthritis with 5 intravenous infusions of 5 mg BT061 every other day with concomitant treatment with 150 mg Diclophenac ( Wijdenes et al., Abstract and poster, EULAR conference, June 2005 ).
  • the antibody described in this study is not disclosed to be suitable for use in lower doses, and it is still desirable to find treatments at lower doses so as to treat a greater number of patients.
  • the present invention provides a pharmaceutical composition for use in treating psoriasis or rheumatoid arthritis comprising a pharmaceutically acceptable carrier and a humanized anti-CD4 antibody capable of activating CD4+CD25+ regulatory T cells, wherein the composition is to be administered to a subject in a dose of the humanized anti-CD4 antibody of 0.2 mg to 5 mg and is to be administered intravenously, wherein the composition is to be administered weekly, every two weeks, every four weeks, every calendar month, or every six weeks, wherein the humanized anti-CD4 antibody comprises an H chain V domain, an L chain V domain, and an IgG1 constant domain, and wherein the H chain V domain is defined by SEQ ID No: 1 and the L chain V domain is defined by SEQ ID No: 2.
  • the invention further provides a pharmaceutical composition for use in treating psoriasis or rheumatoid arthritis in a subject comprising a pharmaceutically acceptable carrier and a humanized anti-CD4 antibody capable of activating CD4+CD25+ regulatory T cells, wherein the subject is to receive a plurality of doses of the composition, wherein the composition is to be administered to the subject in a total dose of the humanized anti-CD4 antibody over a period of 10 days of between 0.2 and 7.5 mg, wherein the composition is to be administered intravenously, wherein the humanized anti-CD4 antibody comprises an H chain V domain, an L chain V domain, and an IgG1 constant domain, and wherein the H chain V domain is defined by SEQ ID No: 1 and the L chain V domain is defined by SEQ ID No: 2.
  • the invention provides a pharmaceutical composition for use in treating psoriasis or rheumatoid arthritis comprising a pharmaceutically acceptable carrier and a humanized anti-CD4 antibody capable of activating CD4+CD25+ regulatory T cells, wherein the composition is to be administered to a subject in a dose of the humanized anti-CD4 antibody of 2 to 20 ⁇ g/kg and is to be administered intravenously, wherein the composition is to be administered weekly, every two weeks, every four weeks, every calendar month, or every six weeks, wherein the humanized anti-CD4 antibody comprises an H chain V domain, an L chain V domain, and an IgG1 constant domain, and wherein the H chain V domain is defined by SEQ ID No: 1 and the L chain V domain is defined by SEQ ID No: 2.
  • the present invention provides a use of a humanized anti-CD4 antibody capable of activating CD4+CD25+ regulatory T cells for the manufacture of a pharmaceutical composition for treating psoriasis or rheumatoid arthritis wherein the composition is to be administered to a subject in a dose of the humanized anti-CD4 antibody of 0.2 mg to 5 mg and is to be administered intravenously, wherein the composition is to be administered weekly, every two weeks, every four weeks, every calendar month, or every six weeks, wherein the humanized anti-CD4 antibody comprises an H chain V domain, an L chain V domain, and an IgG1 constant domain, and wherein the H chain V domain is defined by SEQ ID No: 1 and the L chain V domain is defined by SEQ ID No: 2.
  • the present invention provides a use of a humanized anti-CD4 antibody capable of activating CD4+CD25+ regulatory T cells for the manufacture of a pharmaceutical composition for treating psoriasis or rheumatoid arthritis in a subject, wherein the subject is to receive a plurality of doses of the composition, wherein the composition is to be administered to the subject in a total dose of the humanized anti-CD4 antibody over a period of 10 days of between 0.2 mg and 7.5 mg, wherein the composition is to be administered intravenously, wherein the humanized anti-CD4 antibody comprises an H chain V domain, an L chain V domain, and an IgG1 constant domain, and wherein the H chain V domain is defined by SEQ ID No: 1 and the L chain V domain is defined by SEQ ID No: 2.
  • the present invention provides a use of a humanized anti-CD4 antibody capable of activating CD4+CD25+ regulatory T cells for the manufacture of a pharmaceutical composition for treating psoriasis or rheumatoid arthritis, wherein the composition is to be administered to a subject in a dose of the humanized anti-CD4 antibody of 2 to 20 ⁇ g/kg, wherein the composition is to be administered intravenously, wherein the composition is to be administered weekly, every two weeks, every four weeks, every calendar month, or every six weeks, wherein the humanized anti-CD4 antibody comprises an H chain V domain, an L chain V domain, and an IgG1 constant domain, and wherein the H chain V domain is defined by SEQ ID No: 1 and the L chain V domain is defined by SEQ ID No: 2.
  • low dosages of the antibody BT061 provided effective and specific activation of naturally occurring regulatory T cells (CD4 + CD25 + Tregs) providing an in vivo clinical effect at far lower doses than those previously used, such as those disclosed in WO 2004/083247 .
  • the humanized antibody BT061 did not substantially modulate levels nor induce release of pro-inflammatory cytokines, as compared to other T cell interacting antibodies, for example anti-CD3 antibodies. Further the antibody does not cause a substantial long term depletion of CD4+ lymphocytes.
  • the concentration of the antibody is not especially limited, provided that it is present in a concentration that is low compared to known concentrations.
  • the concentration of the antibody is from 0.1 ⁇ g/ml to 30 mg/ml or, 0.1 to 1000 ⁇ g/ml, and more preferably from 1-500 ⁇ g/ml and 2-250 ⁇ g/ml.
  • the concentration of the antibody is (approximately) any one of 15 ⁇ g/ml, 25 ⁇ g/ml, 125 ⁇ g/ml, 250 ⁇ g/ml, 500 ⁇ g/ml, 1 mg/ml, 12.5 mg/ml or 25 mg/ml.
  • the dosage volume applied to a subject using the composition is not especially limited, provided that it delivers a low overall dosage compared to dosages already known, and is therefore suitable for all patients because of a lower level of side effects and especially in the treatment of individuals who do not tolerate doses as disclosed in WO 2004/083247 .
  • the concentration of the antibody within the dosage volumes can be varied in order to provide the required dosages which are described in this application.
  • the dosage volume will vary depending on the method of administration.
  • the composition is to be administered by intravenous infusion.
  • the dosage volume may be from 0.1 or 0.5 ml up to 500 ml, preferably between 15 and 25 ml, and typically about 20 ml.
  • the composition may be provided in concentrated form and diluted to the strength required for the individuals concerned.
  • the composition is provided in relatively small volumes of about 1, 2, 3, 4 or 5 ml.
  • the composition is provided at the required strength and dosage volume described above (i.e. ready for administration).
  • agents capable of treating autoimmune disease could be administered in the low dosages that are envisaged by the present invention. Whilst known doses of agents capable of treating autoimmune disease are effective in some individuals or disease types, the realisation that it may be effective already in much lower doses has opened up the way for more effective treatment of some autoimmune diseases and classes of patients.
  • the agents that are suitable for use in the present description are those which are capable of activating CD4+CD25+ regulatory T cells.
  • the agent may be a polypeptide, a protein or an antibody. Where the agent is an antibody it may be a monoclonal antibody. Preferably the antibody is a monoclonal anti-CD4 antibody.
  • the antibody may also preferably be an IgG1 antibody and may be an unmodified IgG1 antibody.
  • the agent does not cause a substantial increase in the level of pro-inflammatory cytokines in the subject's blood plasma after administration as compared to the levels seen after administration of anti-CD3 antibodies.
  • the levels of IFN- ⁇ , TNF- ⁇ , IL-6 and/or IL-2 after administration of the agent are not substantially raised compared to plasma levels measured in healthy subjects (see Table A1).
  • the ULN for a specific cytokine given in Table A1 is taken as X then within 96 hours after administration of the agent there may be less than a 20 fold increase in X.
  • cytokine levels prior to administration of the agent are already higher than those observed in healthy subjects (ULN given in Table A1) e.g. due to a modified activation status of immune cells compared to the activation status of the cells in healthy subjects.
  • the concentration for a specific cytokine directly prior to administration of the agent is taken as X and within 96 hours after administration of the agent there may be less than a 20 fold increase in X. Preferably there may be less than a 10 fold increase in X. More preferably these levels are during the period of 10 minutes after the start of administration to 96 hours after the completion of administration.
  • Table A1 Cytokine levels measured in plasma of healthy volunteers. The ULN (upper limit of normal) is calculated based on mean values measured in 39 individual subjects + 2 x standard deviation.
  • the agent does not cause a substantial long-lasting decrease in the cell count of CD4+ lymphocytes in the subject's blood plasma.
  • the cell count of CD4+ lymphocytes in the subject's blood plasma may be above 250 cells/ ⁇ l (or at least 250 cells/ ⁇ l).
  • cytokine and CD4+ lymphocyte effects described above are seen in at least 80% of patients treated.
  • the agent is an unmodified IgG1 antibody, i.e. an antibody which does not include an Fc mutation, and has not been subject to deglycosylation, glycomodification or glycoengineering to reduce Fc receptor interactions, or a fragment or a derivative thereof.
  • the antibodies which are most suitable for use in the present description are humanized anti-CD4 antibodies, or fragments or derivatives thereof, which are capable of activating CD4+CD25+ regulatory T cells. Examples of antibodies which are capable of activating CD4+CD25+ regulatory T cells are discussed in Becker et al., (European Journal of Immunology (2007), Vol. 37: pages 1217-1223 ).
  • the antibody used in the description comprises one or more variable domains which are capable binding to CD4.
  • the antibody may comprise a human constant region (Fc).
  • This constant region can be selected among constant domains from any class of immunoglobulins, including IgM, IgG, IgD, IgA and IgE, and any isotype, including IgG1, IgG2, IgG3 and IgG4.
  • Preferred constant regions are selected among constant domains of IgG, in particular IgG1.
  • the present description also includes any fragment of the antibody comprising the V regions thereof. This comprises in particular Fab, Fab', F(ab)' 2 , Fv and scFv fragments.
  • the antibody is a humanized anti-CD4 antibody or fragment or derivative thereof derived from the mouse monoclonal anti-CD4 antibody B-F5.
  • An example of such an antibody is the BT061 antibody.
  • the humanized antibody BT061 (hB-F5) is derived from mouse B-F5 mAb, and has V domains defined by the following polypeptide sequences:
  • Derivatives of this antibody are also suitable for use in the present descriptoin.
  • Derivatives include those with V domains defined by polypeptide sequences having at least 80%, preferably at least 90%, most preferably at least 95% sequence identity with SEQ ID NO: 1 or SEQ ID NO: 2.
  • Particularly preferred antibodies are those which comprise the complementarity-determining regions (CDRs) of the mouse B-F5 mAb, and retain the ability of hB-F5 to activate CD4+ CD25+ regulatory T cells.
  • CDRs complementarity-determining regions
  • the location of the CDRs within the V H and V K domains is shown in Figures 20 and 21 .
  • Such antibodies can optionally have variations in the sequence of the CDRs that do not substantially affect the specificity and/or affinity of binding.
  • the hB-F5 antibody used further comprises a human constant region (Fc).
  • this constant region can be selected among constant domains from any class of immunoglobulins, including IgM, IgG, IgD, IgA and IgE, and any isotype, including IgG1, IgG2, IgG3 and IgG4.
  • Preferred constant regions are selected among constant domains of IgG, in particular IgG1.
  • the present description also includes any fragment of the hB-F5 antibody or derivative thereof comprising the V regions thereof. This comprises in particular Fab, Fab', F(ab)' 2 , Fv and scFv fragments.
  • a polynucleotide encoding the V domain of the H chain or of the L chain of a BT061 antibody may be fused with a polynucleotide coding for the constant region of a human H or L chain, for the purpose of expressing the complete H and L chains obtained in this way; a sequence coding a signal peptide allowing the secretion of the protein can also be added.
  • the description also makes use of expression cassettes wherein a polynucleotide as described above is linked to appropriate control sequences allowing the regulation of its transcription and translation in a chosen host cell, and recombinant vectors comprising a polynucleotide or an expression cassette of the description.
  • recombinant DNA constructs can be obtained and introduced in host cells by the well-known techniques of recombinant DNA and genetic engineering.
  • the description also makes use of a host cell, transformed by a polynucleotide of the description.
  • a host cell transformed by a polynucleotide of the description.
  • Useful host-cells within the framework of the present description can be prokaryotic or eukaryotic cells.
  • suitable eukaryotic cells one will mention, by way of example, plant cells, cells of yeasts such as Saccharomyces , cells of insects such as Drosophila , or Spodoptera, and mammal cells such as HeLa, CHO, 3T3, C127, BHK, COS, etc.
  • the BT061 (hB-F5) antibody used in the invention can be obtained by culturing a host cell containing an expression vector comprising a nucleic acid sequence encoding said antibody, under conditions suitable for the expression thereof, and recovering said antibody from the host cell culture.
  • DNA sequences encoding mouse B-F5 V H and V K regions are respectively shown in Figure 3 and Figure 4 and under sequence identifiers SEQ ID NO:5 and SEQ ID NO:6.
  • the human V H and V K on which the mouse CDRs are grafted were selected by searching databases for human V H most like the original mouse B-F5 V H and V K .
  • V H region of a human antibody (M26; Accession Number A36006) had the highest homology with B-F5 V H .
  • VK region of another human antibody FK-001; NAKATANI et al., Biotechnology, 7 (1989), 805-810 ) had the highest homology with B-F5 V K .
  • V K Two types of V K differing between them in that the 4 th residue was Leucine or Methionine were constructed and designated as L4L and L4M.
  • VH Two types of VH differing between them in that the 37 th amino acid residue was Leucine or Valine, were constructed and designated as H37L and H37V.
  • the alignment of the polypeptide sequences of B-F5, M26, H37L, and H37V is shown in Figure 21 .
  • the FR residues previously reported to be important for the packing of CDRs Chothia et al., Nature 342(1989), 877 ; Foote et al., J. Mol. Biol., 224(1992), 487 ) are boxed.
  • V H and V K By combining these V H and V K , 4 versions of V regions were designed.
  • expression plasmids for the H chain V H humanized region fused to the constant region of a human y-1 chain ( TAKAHASHI et al., Cell, 29 (1982), 671-679 )
  • L chain VK humanized region fused to the constant region of FK-001 K chain
  • Figure 5 and 6 respectively show the fragments of the plasmids encoding the VH and V K regions of humanized BF-5.
  • Culture supernatants of transfectomas producing the four versions of hB-F5 were collected, and concentrated.
  • the different antibodies were purified from culture supernatants by affinity chromatography using protein A Sepharose and assessed for their CD4 binding activity by measuring, by means of competitive ELISA, their inhibitory activities against the binding of biotinylated mB-F5 to soluble CD4 coated on microtiter plates. Incubation, time is 2 hours for 37°C and overnight for 4°C.
  • H37L/L4L and H37L/L4M were chosen for evaluation.
  • mice B-F5 and humanized B-F5s H37L/L4M IgG1 and H37L/L4L IgG 1 were evaluated.
  • Humanized B-F5s of IgG2 type H37L/L4M IgG2 and H37L/L4L IgG2 were also tested.
  • PBMCs peripheral blood mononuclear cells
  • Murine and hB-F5s could moderately inhibit ConA-induced proliferation, but the activities varied from antibody to antibody and/or from donor to donor. Also, murine and hB-F5s were able to inhibit Ag-specific PBMC proliferation induced by PPD.
  • IgG1 type of hB-F5 inhibited PPD-induced proliferation more effectively (as high as 70% inhibition) than mB-F5.
  • IgG1 type seemed to be more effective than IgG2 type of which inhibitory activity was almost the same as mB-F5.
  • H37L/L4M was more effective than H37L/L4L.
  • IgG2 type of H37L/L4M and H37L/L4L had almost the same inhibitory activities.
  • H37L/L4M IgG1 was chosen for further evaluation, and it is this antibody which is named BT061 and is employed to demonstrate the present invention in the Examples provided in this application.
  • the pharmaceutical composition and medicaments used in the present description are preferably capable of treating an autoimmune disease in patients benefiting from lower doses.
  • Such patients include all patients because of a lower level of side effects but especially individuals who do not tolerate doses as disclosed in WO 2004/08324 .
  • the present description also provides use of a humanized anti-CD4 antibody or fragment or derivative thereof in the manufacture of a medicament effective against an autoimmune disease, wherein the humanised antibody capable of activating CD4+CD25+ regulatory T cells, and wherein the medicament comprises the antibody in a concentration of from 10 ⁇ g/ml to 150 mg/ml, preferably from 0.5 mg/ml to 75 mg/ml.
  • the description further provides use of a humanized anti-CD4 antibody or fragment or derivative thereof in the manufacture of a medicament effective against an autoimmune disease, wherein the humanised antibody is capable of activating CD4+CD25+ regulatory T cells, and wherein the medicament is administered to a subject in a single dose or a plurality of doses in an amount of the antibody of from 0.2 to 30 mg per dose.
  • the present description also provides a pharmaceutical composition for treating an autoimmune disease comprising a pharmaceutically acceptable carrier and an agent capable of activating CD4+CD25+ regulatory T cells, wherein the composition is to be administered to a subject in a dose of the agent from 0.2 mg to 30 mg, 0.2 to 20 mg per dose, 0.3 mg to 7.5 mg, 0.3 to 5 mg per dose or preferably 0.3 to 1 mg per dose. Most preferably the range of milligrams per dose extends between 0.3 mg to 0.9 mg, or 0.3 to 0.99 mg.
  • the subject is to receive a plurality of doses.
  • dosage over a period of 10 days is between 0.2 to less than 25 mg, more preferably between 0.2 and 20 mg and most preferably between 0.2 to less than 10 mg.
  • the dosage over a period of 5 days should be between 0.2 to less than 15 mg, preferably between 0.2 and 12 mg, and more preferably between 0.2 to less than 5 mg.
  • the dosages over a period of 10 days are between 0.2 to less than 10 mg, most preferably between 0.2 and 7.5 mg.
  • the dosages over a period of 10 days are between 1 mg to 30 mg, more preferably between 5 mg and 30 mg.
  • the dose can also be calculated on the basis of the body surface area (BSA) of the subject.
  • the dose of the agent to the subject is from 0.10 to 20 mg/m 2 body surface area of the patient, preferably from 0.12 to 15 mg/m 2 , more preferably 0.20 to 10 mg/m 2 and most preferably 0.30 to 0.50 mg/m 2 .
  • the dose can be calculated based on the body weight of the subject.
  • the dose of the agent to the subject is from 1 to 500 ⁇ g/kg, preferably 2 to 400 ⁇ g/kg, more preferably 2 to 250 ⁇ g/kg and most preferably 2.5 to 20 ⁇ g/kg.
  • the dosages over a period of 10 days are between 0.20 to 10 mg/m 2 , more preferably between 0.20 to 4 mg/m 2 , or between 2 to 250 ⁇ g/kg, more preferably between 2 to 100 ⁇ g/kg.
  • the dosages over a period of 10 days are between 0.30 to 20 mg/m 2 , more preferably between 0.5 to 20 mg/m 2 , or between 2.5 to 500 ⁇ g/kg more preferably between 20 to 500 ⁇ g/kg.
  • the frequency of administration is not especially limited, provided that it does not interfere with the effectiveness of the treatment. It is preferred that the plurality of doses are administered on at least the following bases: daily, every other day, weekly, every 4 weeks, every 6 weeks, every 12 weeks, every 24 weeks, every calendar month, every 6 calendar months or yearly.
  • the doses may be separated by at least one day, or alternatively by at least one week, or by at least one month or by at least 3 months or by at least 6 months or by at least one year (meaning that the doses are taken every day or every week, or every month or every 6 months or every year).
  • the plurality of doses are taken from every 1 to 31 days, or every 1-12 months.
  • the length of treatment is not especially limited, and typically in treatment of autoimmune diseases, the treatment proceeds indefinitely, or until symptoms are reduced to a manageable level for the patient. Generally the dose is administered to the subject for at least 1 month.
  • kits for a use as defined above wherein the kit comprises a plurality of medicament dosages for simultaneous, sequential or separate administration to a subject.
  • Also provided is a method of treatment of an autoimmune disease which method comprises administering a medicament to a subject, wherein the medicament comprises an agent capable of activating CD4+CD25+ regulatory T cells, and wherein the medicament is administered to the subject in an amount as described above.
  • the agent is a humanized anti-CD4 antibody or fragment or derivative thereof derived from the mouse monoclonal anti-CD4 antibody B-F5
  • the pharmaceutical composition and medicaments used according to the present description are for treating an autoimmune disease.
  • the autoimmune disease is selected from psoriasis, rheumatoid arthritis, multiple sclerosis, type-1 diabetes, inflammatory bowel diseases, Crohn's disease, autoimmune thyreoditis, autoimmune myasthenia gravis, systemic lupus erythematosus, ulcerative colitis, atopic dermatitis, myocarditis and transplantation-related diseases such as graft-versus-host or host-versus-graft reactions, or general organ tolerance issues.
  • the autoimmune disease is psoriasis.
  • Psoriasis is a disorder which causes psoriatic lesions or plaques on the sufferer's skin.
  • PASI Psoriasis Area and Severity Index
  • PASI 0.1 E h + I h + D h A h + 0.2 E u + I u + D u A u + 0.3 E t + I t + D t A t + 0.4 E l + I l + D l A t PASI score ranges from 0-72. A score of 0 means no psoriasis, while a score of 72 represents the most severe psoriasis.
  • the pharmaceutical composition of the present description is capable of treating psoriasis by providing at least a 40 %, and preferably at least a 50 %, improvement in the PASI score of the patient.
  • the subject has a PASI score of at least 10 prior to treatment.
  • the pharmaceutical composition is to be administered intravenously, subcutaneously or intramuscularly in the dosages specified herein.
  • the dose is between 0.2 mg to 7.5 mg, more preferably between 0.3 to 5 mg.
  • the dosage over a period of 10 days is preferably between 0.2 to less than 10 mg.
  • the dose is to be administered subcutaneously or intramuscularly it is preferred that the dose is between 0.2 mg to 30 mg, more preferably between 5 mg to 30 mg.
  • the dosage over a period of 10 days is preferably between 0.2 to less than 25 mg.
  • compositions are for treating rheumatoid arthritis.
  • Rheumatoid arthritis is an autoimmune disease which causes chronic inflammation of joints and surrounding tissues, and can also affect other tissues and body organs.
  • ACR American College of Rheumatology
  • CRP C-reactive protein
  • compositions for treating rheumatoid arthritis are preferably to be administered intravenously, subcutaneously or intramuscularly in the dosages specified herein.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • Secondary treatment of arthritis includes corticosteroids (e.g. prednisone and dexamethasone), slow acting antirheumatic drugs (SAARDs) or disease-modifying antirheumatic drugs (DMARDs), e.g., methotrexate, penicillinamine, cyclophosphamide, gold salts, azothipoprine, leflunomide, etc.
  • Corticosteroids the synthetic versions of the body's cortisone hormone, are used to inhibit RA progression (e.g. prednisone and dexamethasone).
  • BRMs biological-response modifiers
  • compositions are to be administered in combination with drugs currently used to treat rheumatoid arthritis.
  • the compositions are to be administered with one of the drugs mentioned above, preferably methotrexate.
  • Known drugs such as methotrexate
  • the pharmaceutical composition of the present invention can be administered simultaneously, sequentially or separately.
  • Human PBMCs were isolated by density gradient centrifugation and stained with FITC-labelled anti-CD3 antibody (345763; Becton/Dickinson) and serial dilutions of BT061.
  • BT061 binding was detected with an phycoerythrin labelled human IgG antibody (109-116-098; Jackson, Immunoresearch).
  • flow cytometric analysis the mean fluorescence intensity of CD3+ BT061 binding lymphocytes was determined.
  • BT061 binds to human lymphocytes at low concentrations. Below 10 ng/ml the half maximal saturation of binding is observed. Saturation is found at 100 ng/ml. The concentrations are as expected for patients which receive doses of 30 and 300 ⁇ g.
  • EXAMPLE 2 Inhibition of proliferation of CD8+ T-cells by BT061-stimulated T reg cells (results shown in figure 7)
  • CD25 high Tregs were separated from buffy coats and/or leukapheresis of healthy volunteers by magnetic bead cell separation according to the following protocol.
  • CD4 + CD25 + regulatory T cells were isolated from buffy coats of healthy volunteers by 2 steps.
  • CD4 + T cells using 2-4 ⁇ l CD4-MACS-Multisort-Beads (Miltenyi Biotec) per 10 7 PBMCs were positively selected. After 15 minutes of incubation, magnetic selection was performed.
  • positively isolated cells were depleted of CD25-expressing non-CD4 cells with CD8-, CD19- and CD14-Dynabeads (Dynal, Oslo, Norway). The resulting CD4 + CD25 + T cells were 95-98% pure.
  • Untouched CD4 + CD25 - T cells were isolated by negative selection from PBMC by depleting CD8, CD19, CD56, CD14, CD235a, CD25 and CD45RO expressing cells with Dynabeads.
  • the purification scheme is shown in Figure 2 .
  • BT061 reproducibly induces suppressive activity in Tregs in a dose-dependent manner, resulting in inhibition of the proliferation of alloreactive CD8+ T cells. BT061 stimulates CD4+CD25+ Tregs which directly inhibit CD8+ T cells.
  • results confirm that there is inhibition of the proliferation of CD8+ T cells at concentrations as low as 10ng/ml corresponding to low dose application in patients of 30 ⁇ g.
  • hB-F5 BT061 The ability of hB-F5 BT061 to treat an autoimmune disease is being tested on 56 patients suffering from moderate to severe chronic psoriasis.
  • the trial comprises a single dose escalation study to assess the safety and efficacy of hB-F5.
  • the antibody/placebo is to be infused in the forearm vein according to medically accepted procedures.
  • the total volume is administered as a single continuous intravenous infusion over a period of 2 hours via a perfusor (Fresenius Pilot C, Fresenius AG, Germany).
  • Each dose of the antibody is diluted with a 0.9% sodium chloride injection (B. Braun Melsungen AG, Germany) up to a total volume of 20 ml.
  • the antibody is to be administered as a single subcutaneous injection.
  • the same procedure applies for the placebo.
  • PASI Psoriasis Area and Severity Index
  • the patient's PASI score is assessed before the trial to provide a "baseline" value at day 0, and repeatedly during the trial at days 5, 7, 14, 21, 28, 42, 56 and 75.
  • the PASI scores for the patients in dose group I are shown in Table C together with the percentage improvement in the PASI score from the baseline.
  • the PASI scores for the patients in dose group II are shown in Table D1 together with the percentage improvement in the PASI score from the baseline.
  • the PASI scores for the patients in dose group III are shown in Table D2 together with the percentage improvement in the PASI score from the baseline.
  • PASI scores for the patients in dose group IV are shown in Table D3 together with the percentage improvement in the PASI score from the baseline. TABLE C - PASI scores for the patients in dose group I (0.5mg intravenous dose) over course of trial Patient 1 2 3 4 5 6 7 8 Rel.
  • the PASI scores against time for individual patients are shown in graph form in Figures 8A to 8H and in Figures 9A to 9H .
  • the graphs shown in Figures 8A to 8H represent PASI scores for patients from dose group I, while the graphs shown in Figures 9A to 9H represent PASI scores for patients from dose group II.
  • Patients in dose group III also show an improvement in their PASI score, with six out of eight patients showing a greater than 20% improvement and two of those six showing a greater than 30% improvement after treatment. However, the improvement was not as significant as that seen in patients from dose group I and dose group II which received a lower dose of the antibody. Some efficacy is also seen in the patients of dose group IV. In particular patients 1, 4, 5 and 8 in this dose group (as shown in Table D3) show a clear improvement in their PASI scores, although this is limited in comparison to patients of dose groups I to III.
  • Figures 14A and 14B provide photographic evidence of the improvement in the level of psoriasis before and after treatment.
  • Figure 14A shows an area of psoriasis on the skin of a patient in dose group II prior to administration.
  • Figure 14B shows the same area of psoriasis 28 days after administration. The areas of improvement are marked on Figure 14B with black boxes.
  • BT061 provides effective treatment of moderate and severe chronic psoriasis even with a dose as low as 0.5mg. Further, the single dose provides a therapeutic effect which can still be seen six to eight weeks afterwards.
  • BT061 Thirty volunteers received BT061 by intravenous administration in 10 dosage groups, with 3 volunteers per group. Further, 15 volunteers received BT061 by subcutaneous administration in 5 dosage groups also with 3 volunteers per group.
  • the administration of BT061 intravenously is illustrated Table F below: TABLE F - Intravenous dose of BT061 Administration of BT061 Total dose of BT061 mab Volume of BT061-12.5 mg Volume of BT061-25 mg Volume of BT061-50 mg 3.5 ⁇ g 0.28 ⁇ l - - 20 ⁇ g 1.6 ⁇ l - - 100 ⁇ g 8 ⁇ l - - 500 ⁇ g 40 ⁇ l - - 2.5 mg 0.2 ml - - 5 mg 0.4 ml - - 10 mg 0.8 ml - - 20 mg - 0.8 ml - 40 mg - - 0.8 ml 60 mg 0.8 ml - 1 ml
  • Each dose is diluted with 0.9% sodium chloride injection up to a total volume of 20 ml.
  • the dose is administered as a single continuous intravenous infusion over 2 hours.
  • BT061 subcutaneously is illustrated in Table G below: TABLE G - Subcutaneous dose of BT061 Administration of BT061 Total dose of BT061 mab Volume of BT061-12.5 mg Volume of BT061-25 mg Volume of BT061-50 mg 5 mg 0.4 ml - - 10 mg 0.8 ml - - 20 mg - 0.8 ml - 40 mg - - 0.8 ml 60 mg - - 1 ml + 0.2 ml
  • Each dose is injected as a single bolus injection.
  • the volunteers were assessed over a period of 3 months after the injection.
  • plasma samples were taken before administration and at 3, 6, 12, 24, 36, 48, 56, 72, 88, 96, 120, 144 and 168 hours after administration and on day 75.
  • plasma samples were taken before administration and at 30 minutes, 1, 2, 3, 6, 12, 24, 36, 48, 72, 96, 120, 144 and 168 hours after administration.
  • the plasma samples were analyzed using standard ELISA methodology to establish cytokine levels.
  • the relevant cytokines analyzed included: IFN- ⁇ , TNF- ⁇ , IL-6 and IL-2.
  • the plasma samples were also analyzed using standard methods of flow cytometry to measure the number of CD4+ lymphocytes.
  • Induction of cytokine release is a common immediate complication occurring with the use of T cell interacting therapeutic antibodies, such as ATG, OKT3, CAMPATH-1H and humanized anti-CD3 mAbs (TRX4, Visilizumab and Teplizumab).
  • T cell interacting therapeutic antibodies such as ATG, OKT3, CAMPATH-1H and humanized anti-CD3 mAbs (TRX4, Visilizumab and Teplizumab).
  • TRX4 Visilizumab and Teplizumab
  • the symptoms mainly include moderate fever, headaches and self-limiting gastrointestinal manifestations.
  • Side effects correlated with cytokine induction after antibody administration require the application of additional drugs such as the antihistamine diphenhydramine hydrochloride and/or the anti-inflammatory agent ibuprofen.
  • OKT3 muromonab-CD3
  • a murine CD3 specific therapeutic monoclonal antibody there have even been deaths reported, and severe side effects limit the clinical use of this antibody mainly to immunosuppressed patients.
  • humanized FcR-non-binding CD3-specific monoclonal antibodies that are presently used in the clinic for the treatment of autoimmune disease (Teplizumab and TRX4) exhibit reduced side effects induced by T-cell activation and/or by activation of Fc receptor expressing cells after the first dose, as compared with FcR-binding CD3-specific antibodies such as OKT3, some degree of T-cell activation and activation of Fc receptor expressing cells is still observed that leads to cytokine release generally connected to cytokine dependent side effects.
  • cytokine induction observed in healthy volunteers after intravenous or subcutaneous application of BT061 was comparably low and transient as compared to anti-CD3 antibodies. Cytokine induction generally increased with increasing dosage. However, even at the highest doses of 40 to 60 mg cytokine induction is much lower than that seen with other T cell interacting monoclonal antibodies.
  • the median peak concentration for each cytokine is calculated as follows: The median of the highest cytokine concentrations observed after administration of the antibody.
  • Figure 10A and B show the TNF ⁇ and IL-6 release observed in healthy volunteers after intravenous or subcutaneous administration of BT061 in comparison to those released after administration of anti-CD3 monoclonal antibodies, Teplizumab and TRX4.
  • the normal values of these cytokines were taken from Straub et al., (2007, Arthr. & Rheumat.).
  • Figure 11 shows the IL-2 and IFN- ⁇ plasma levels after administration of intravenous or subcutaneous BT061.
  • the median peak levels were calculated from the 40 and 60 mg dose group measured within 4 days after antibody injection.
  • BT061 induced only marginal and transient cytokine release. TNF- ⁇ and IL-6 levels were slightly increased.
  • Figure 10A and B shows that the median peak values of IL-6 and TNF ⁇ cytokine levels detected in plasma after application of BT061 (40 and 60 mg) are lower than those seen after treatment with the CD3 specific therapeutic antibodies Teplizumab and TRX4.
  • BT061 did not lead to substantially increased levels of IFN- ⁇ and IL-2 ( Figure 11 ) as was reported for the application of TRX4 ( Keymeulen et al., 2005 N. Engl. J. Med. Type 1 Diabetes patients).
  • the trial also included a study of the numbers of CD4-positive lymphocytes in plasma samples collected.
  • Figure 12 shows the CD4 cell counts (cells per ml plasma) in volunteers treated with the single intravenous dose of BT061.
  • the data points represent the mean values of the 3 patients in each dose group.
  • Dotted lines indicate the upper limit of normal (ULN) and the lower limit of normal (LLN).
  • the ULN and the LLN were calculated based on cell counts measured in 11 healthy volunteers using identical methodology to that used to measure the cell counts in those volunteers receiving BT061.
  • the ULN and the LLN represent the mean of all the 11 healthy volunteer values + (or -) the standard deviation.
  • Norm values for CD4 cell counts were calculated to be between 443 CD4 cells per ⁇ l (lower limit of normal; LLN) and 1324 CD4 cells per ⁇ l (upper limit of normal; ULN).
  • Figure 13 shows the CD4 cell counts (cells per ml plasma) in volunteers treated with the single subcutaneous dose of BT061. As with Figure 12 , the data points represent the mean values of the 3 patients in each dose group. Dotted lines indicate the upper limit of normal (ULN) and the lower limit of normal (LLN).
  • CD4 specific monoclonal antibodies known in the art (such as those reviewed in Strand et al., 2007) achieve immuno-suppression via CD4-positive lymphocyte depletion.
  • the drawback of these antibodies is that treated individuals become immuno-compromised, and are susceptible to other infections.
  • CD4 cell counts in four volunteers of the intravenous dose groups showed CD4 levels that were below these norm values as follows: 1 volunteer of the 100 ⁇ g intravenous dose: 400 CD4 cells per ⁇ l; 1 volunteer of the 5 mg group: 419 CD4 cells per ⁇ l; 1 volunteer of the 10 mg group: 440 CD4 cells per ⁇ l; and 1 volunteer of the 20 mg group: 392 CD4 cells per ⁇ l.
  • CD4 cell counts in the remaining 26 volunteers of the intravenous dose groups were within the norm values 72 hours after administration of BT061.
  • BT061 in contrast to depleting CD4 specific mAbs, BT061 only induced a transient decline of CD4-positive cells followed by a general recovery. From the transient decline and rapid general recovery to norm values it is concluded that a transient redistribution of the CD4-positive cells has taken place, rather than depletion of these cells.
  • the ability of hB-F5 BT061 to treat rheumatoid arthritis is being tested on patients suffering from this disease.
  • the trial comprises a multiple dose study involving 96 patients, divided into 12 groups. In each group two patients receive a placebo while 6 patients receive BT061. Patients are dosed once a week over a period of 6 weeks.
  • the subcutaneous dose groups are: 1.25 mg, 6.25 mg. 12. 5 mg, 25 mg, 50 mg, 75 mg and 100 mg.
  • the intravenous dose groups are: 0.5 mg, 2 mg, 6.25 mg, 12.5 mg and 25 mg.
  • the patients are numbered 101, 102, 103, 104, 105, 106, 107 and 108.
  • the patients are numbered 201-208.
  • the patients are numbered 301-308.
  • the patients are numbered 401-408.
  • the patients are numbered 501-508.
  • the 6.25 mg intravenous dose group the patients are numbered 601-608.
  • the intravenous and subcutaneous administration procedure was the same as that described in Example 3 for the psoriasis trial.
  • the level of rheumatoid arthritis is recorded weekly by assessing the ACR parameters and in particular studying the number of tender and swollen joints and following the levels of C-reactive protein (CRP) and the erythrocyte sedimentation rate (ESR). These parameters are assessed before the trial to provide a "baseline” value at day 0, and repeatedly during the trial period and thereafter at 8, 22 and 43 days after the administration period is finished (i.e. follow up (FU) day 8, FU day 22 and FU day 43).
  • CRP C-reactive protein
  • ESR erythrocyte sedimentation rate
  • Tables below provide the data obtained from the trial. Specifically Tables M to S provide the number of tender and swollen joints over the course of the trial.
  • Figure 15 shows the percentage of patients from the dose groups receiving 1.25 mg. 6.25 mg, 12.5 mg and 25 mg subcutaneous BT061 achieving at least a 20% improvement of relevant ACR parameters over the course of the trial, and the percentage of patients achieving at least an ACR20 response at week 7.
  • Figure 16A and 16B show results for the number of tender and swollen joints exhibited by patients from the 25 mg subcutaneous BT061 dose group over a six week period. Several patients exhibit a reduction in the number of tender and swollen joints over a period of the treatment. The results for one responder patient and one non-responder patient from this dose group are shown in Figures 17A and 17B , respectively. The responder shows a significant improvement in the number of tender and swollen joints and in pain levels.
  • Figures 18A, 18B , 19A and 19B show the number of tender joints in the 1.25 mg subcutaneous, 6.25 mg subcutaneous, 50 mg subcutaneous and 6.25 mg intravenous dose groups respectively, over the course of the trial and in the weeks thereafter.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Diabetes (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Endocrinology (AREA)
  • Rheumatology (AREA)
  • Neurology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Emergency Medicine (AREA)
  • Dermatology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Pain & Pain Management (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Transplantation (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Claims (13)

  1. Composition pharmaceutique pour une utilisation dans le traitement du psoriasis ou de la polyarthrite rhumatoïde, comprenant un support pharmaceutiquement acceptable et un anticorps anti-CD4 humanisé capable d'activer les lymphocytes T régulateurs CD4+CD25+, ladite composition étant destinée à être administrée à un sujet à une dose de l'anticorps anti-CD4 humanisé de 0,2 mg à 5 mg et étant destinée à être administrée par voie intraveineuse, ladite composition étant destinée à être administrée de manière hebdomadaire, toutes les deux semaines, toutes les quatre semaines, tous les mois ou toutes les six semaines,
    dans laquelle l'anticorps anti-CD4 humanisé comprend un domaine V de chaîne H, un domaine V de chaîne L et un domaine constant de IgG1,
    et dans laquelle le domaine V de chaîne H est défini par la SEQ ID N° 1 et le domaine V de chaîne L est défini par la SEQ ID N° 2.
  2. Composition pharmaceutique pour une utilisation suivant la revendication 1, ladite composition étant destinée à être administrée à une dose de l'anticorps anti-CD4 humanisé de 0,3 mg à 1 mg.
  3. Composition pharmaceutique pour une utilisation dans le traitement du psoriasis ou de la polyarthrite rhumatoïde chez un sujet, comprenant un support pharmaceutiquement acceptable et un anticorps anti-CD4 humanisé capable d'activer les lymphocytes T régulateurs CD4+ CD25+, le sujet étant destiné à recevoir une pluralité de doses de la composition, la composition étant destinée à être administrée au sujet à une dose totale de l'anticorps anti-CD4 humanisé pendant une période de temps de 10 jours entre 0,2 et 7,5 mg, la composition étant destinée à être administrée par voie intraveineuse,
    dans laquelle l'anticorps anti-CD4 humanisé comprend un domaine V de chaîne H, un domaine V de chaîne L et un domaine constant de IgG1,
    et dans laquelle le domaine V de chaîne H est défini par la SEQ ID N° 1 et le domaine V de chaîne L est défini par la SEQ ID N° 2.
  4. Composition pharmaceutique pour une utilisation dans le traitement du psoriasis ou de la polyarthrite rhumatoïde, comprenant un support pharmaceutiquement acceptable et un anticorps anti-CD4 humanisé capable d'activer les lymphocytes T régulateurs CD4+CD25+, ladite composition étant destinée à être administrée à un sujet à une dose de l'anticorps anti-CD4 humanisé de 2 à 20 µg/kg et étant destinée à être administrée par voie intraveineuse, ladite composition étant destinée à être administrée de manière hebdomadaire, toutes les deux semaines, toutes les quatre semaines, tous les mois ou toutes les six semaines,
    dans laquelle l'anticorps anti-CD4 humanisé comprend un domaine V de chaîne H, un domaine V de chaîne L et un domaine constant de IgG1,
    et dans laquelle le domaine V de chaîne H est défini par la SEQ ID N° 1 et le domaine V de chaîne L est défini par la SEQ ID N° 2.
  5. Composition pharmaceutique pour une utilisation suivant la revendication 4, ladite composition étant destinée à être administrée à une dose de l'anticorps anti-CD4 humanisé de 20 µg/kg.
  6. Composition pharmaceutique pour une utilisation suivant l'une quelconque des revendications précédentes, ladite composition étant fournie en un volume de dose de 0,5 à 500 ml, de préférence de 15 à 25 ml.
  7. Composition pharmaceutique pour une utilisation suivant l'une quelconque des revendications 1, 2, 4 et 5, la dose étant destinée à être administrée une fois par semaine, une fois toutes les deux semaines ou une fois toutes les quatre semaines.
  8. Composition pharmaceutique pour une utilisation suivant l'une quelconque des revendications 1, 2, 4 et 5, la dose étant destinée à être administrée de manière hebdomadaire ou mensuelle.
  9. Utilisation d'un anticorps anti-CD4 humanisé capable d'activer les lymphocytes T régulateurs CD4+ CD25+ pour la production d'une composition pharmaceutique pour le traitement du psoriasis ou de la polyarthrite rhumatoïde, dans laquelle la composition ést destinée à être administrée à un sujet à une dose de l'anticorps anti-CD4 humanisé de 0,2 mg à 5 mg et est destinée à être administrée par voie intraveineuse, dans laquelle la composition est destinée à être administrée de manière hebdomadaire, toutes les deux semaines, toutes les quatre semaines, tous les mois ou toutes les six semaines,
    dans laquelle l'anticorps anti-CD4 humanisé comprend un domaine V de chaîne H, un domaine V de chaîne L et un domaine constant de IgG1,
    et dans laquelle le domaine V de chaîne H est défini par la SEQ ID N° 1 et le domaine V de chaîne L est défini par la SEQ ID N° 2.
  10. Utilisation d'un anticorps anti-CD4 humanisé capable d'activer les lymphocytes T régulateurs CD4+CD25+ pour la production d'une composition pharmaceutique pour le traitement du psoriasis ou de la polyarthrite rhumatoïde chez un sujet, dans laquelle le sujet est destiné à recevoir une pluralité de doses de la composition, dans laquelle la composition est destinée à être administrée au sujet à une dose totale de l'anticorps anti-CD4 humanisé pendant une période de temps de 10 jours entre 0,2 mg et 7,5 mg, dans laquelle la composition est destinée à être administrée par voie intraveineuse,
    dans laquelle l'anticorps anti-CD4 humanisé comprend un domaine V de chaîne H, un domaine V de chaîne L et un domaine constant de IgG1,
    et dans laquelle le domaine V de chaîne H est défini par la SEQ ID N° 1 et le domaine V de chaîne L est défini par la SEQ ID N° 2.
  11. Utilisation d'un anticorps anti-CD4 humanisé capable d'activer les lymphocytes T régulateurs CD4+CD25+ pour la production d'une composition pharmaceutique pour le traitement du psoriasis ou de la polyarthrite rhumatoïde, dans laquelle la composition est destinée à être administrée à un sujet à une dose de l'anticorps anti-CD4 humanisé de 2 à 20 µg/kg, dans laquelle la composition est destinée à être administrée par voie intraveineuse, dans laquelle la composition est destinée à être administrée de manière hebdomadaire, toutes les deux semaines, toutes les quatre semaines, tous les mois ou toutes les six semaines,
    dans laquelle l'anticorps anti-CD4 humanisé comprend un domaine V de chaîne H, un domaine V de chaîne L et un domaine constant de IgG1,
    et dans laquelle le domaine V de chaîne H est défini par la SEQ ID N° 1 et le domaine V de chaîne L est défini par la SEQ ID N° 2.
  12. Utilisation suivant la revendication 9 ou la revendication 11, dans laquelle la dose est destinée à être administrée : (i) une fois par semaine, une fois toutes les deux semaines ou une fois toutes les quatre semaines ; ou (ii) de manière hebdomadaire ou mensuelle.
  13. Utilisation suivant l'une quelconque des revendications 9 à 12, dans laquelle la composition est fournie en un volume de dose de 0,5 à 500 ml, de préférence de 15 à 25 ml.
EP09727585.3A 2008-03-13 2009-03-10 Schema posologique pour traiter le psoriasis et l'arhtrite rhumatoide Not-in-force EP2265643B1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GB0804686A GB0804686D0 (en) 2008-03-13 2008-03-13 Humanized antibody
GB0817811A GB0817811D0 (en) 2008-09-29 2008-09-29 Agent for treating disease
PCT/EP2009/052809 WO2009121690A1 (fr) 2008-03-13 2009-03-10 Agent pour traiter une maladie

Publications (2)

Publication Number Publication Date
EP2265643A1 EP2265643A1 (fr) 2010-12-29
EP2265643B1 true EP2265643B1 (fr) 2016-10-19

Family

ID=40756949

Family Applications (1)

Application Number Title Priority Date Filing Date
EP09727585.3A Not-in-force EP2265643B1 (fr) 2008-03-13 2009-03-10 Schema posologique pour traiter le psoriasis et l'arhtrite rhumatoide

Country Status (16)

Country Link
US (1) US9334325B2 (fr)
EP (1) EP2265643B1 (fr)
JP (1) JP5597553B2 (fr)
KR (1) KR20100135257A (fr)
CN (1) CN102027017A (fr)
AU (1) AU2009231325B2 (fr)
BR (1) BRPI0909048A2 (fr)
CA (1) CA2718184A1 (fr)
CO (1) CO6311007A2 (fr)
CR (1) CR11724A (fr)
IL (1) IL207891A0 (fr)
MX (1) MX2010010028A (fr)
RU (1) RU2539110C2 (fr)
SG (1) SG190598A1 (fr)
WO (1) WO2009121690A1 (fr)
ZA (1) ZA201007215B (fr)

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7832566B2 (en) 2002-05-24 2010-11-16 Biomet Biologics, Llc Method and apparatus for separating and concentrating a component from a multi-component material including macroparticles
US20060278588A1 (en) 2002-05-24 2006-12-14 Woodell-May Jennifer E Apparatus and method for separating and concentrating fluids containing multiple components
EP1460088A1 (fr) 2003-03-21 2004-09-22 Biotest AG Anticorps humanisé contre CD4 avec des caractéristiques immunosuppressives
US8034014B2 (en) 2007-03-06 2011-10-11 Biomet Biologics, Llc Angiogenesis initation and growth
US20080269762A1 (en) * 2007-04-25 2008-10-30 Biomet Manufacturing Corp. Method and device for repair of cartilage defects
WO2009108890A1 (fr) 2008-02-27 2009-09-03 Biomet Biologics, Llc Procédés et compositions pour administrer un antagoniste de récepteur d’interleukine-1 antagonist
US8753690B2 (en) * 2008-02-27 2014-06-17 Biomet Biologics, Llc Methods and compositions for delivering interleukin-1 receptor antagonist
BRPI0909180A2 (pt) * 2008-03-13 2016-08-09 Biotest Ag composição farmacêutica, e, método de tratamento de uma doença autoimune
ES2610327T3 (es) * 2008-03-13 2017-04-27 Biotest Ag Régimen de dosificación de anticuerpos anti-CD4 para el tratamiento de enfermedades autoinmunitarias
MX2011003335A (es) 2008-09-29 2011-04-27 Biotest Ag Composicion para tratar enfermedad.
JP5844258B2 (ja) 2009-08-27 2016-01-13 バイオメット、バイオロジクス、リミテッド、ライアビリティー、カンパニーBiomet Biologics, Llc インターロイキン−1受容体アンタゴニストの生産のための植込み型装置
US20110052561A1 (en) * 2009-08-27 2011-03-03 Biomet Biologics,LLC Osteolysis treatment
GB0920944D0 (en) 2009-11-30 2010-01-13 Biotest Ag Agents for treating disease
WO2012030593A2 (fr) 2010-09-03 2012-03-08 Biomet Biologics, Llc Méthodes et compositions permettant d'administrer un antagoniste du récepteur de l'interleukine 1
US9011846B2 (en) 2011-05-02 2015-04-21 Biomet Biologics, Llc Thrombin isolated from blood and blood fractions
US10143725B2 (en) 2013-03-15 2018-12-04 Biomet Biologics, Llc Treatment of pain using protein solutions
US9950035B2 (en) 2013-03-15 2018-04-24 Biomet Biologics, Llc Methods and non-immunogenic compositions for treating inflammatory disorders
US9895418B2 (en) 2013-03-15 2018-02-20 Biomet Biologics, Llc Treatment of peripheral vascular disease using protein solutions
US10208095B2 (en) 2013-03-15 2019-02-19 Biomet Manufacturing, Llc Methods for making cytokine compositions from tissues using non-centrifugal methods
US9878011B2 (en) 2013-03-15 2018-01-30 Biomet Biologics, Llc Treatment of inflammatory respiratory disease using biological solutions
US20140271589A1 (en) 2013-03-15 2014-09-18 Biomet Biologics, Llc Treatment of collagen defects using protein solutions
US9758806B2 (en) 2013-03-15 2017-09-12 Biomet Biologics, Llc Acellular compositions for treating inflammatory disorders
EP3074507B1 (fr) 2013-11-26 2022-01-05 Biomet Biologics, LLC Procédés de médiation des phénotypes macrophagiques
EP3338800A1 (fr) * 2014-02-21 2018-06-27 IDAC Theranostics, Inc. Agent thérapeutique pour un cancer solide
US10441635B2 (en) 2014-11-10 2019-10-15 Biomet Biologics, Llc Methods of treating pain using protein solutions
US9763800B2 (en) 2015-03-18 2017-09-19 Biomet C. V. Implant configured for hammertoe and small bone fixation

Family Cites Families (73)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT90657B (pt) 1988-05-27 1995-03-01 Ortho Pharma Corp Processo para a preparacao de peptidos que bloqueiam a ligacao de hiv-1 a proteina cd4 receptora
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
AU5656890A (en) 1989-05-02 1990-11-29 Genelabs Incorporated Chemically modified cd4 peptide fragments having anti-retroviral properties
GB8912497D0 (en) 1989-05-31 1989-07-19 Cobbold Stephen P Monoclonal antibodies
US5690933A (en) 1989-05-31 1997-11-25 Glaxo Wellcome Inc. Monoclonal antibodies for inducing tolerance
US20020099179A1 (en) 1989-12-21 2002-07-25 Linda K. Jolliffe Cdr-grafted antibodies
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
HUT60768A (en) 1990-03-16 1992-10-28 Sandoz Ag Process for producing cd25 fixing molecules
US7084260B1 (en) 1996-10-10 2006-08-01 Genpharm International, Inc. High affinity human antibodies and human antibodies against human antigens
US5871732A (en) 1990-11-27 1999-02-16 Biogen, Inc. Anti-CD4 antibody homologs useful in prophylaxis and treatment of AIDS, ARC and HIV infection
US7192584B2 (en) 1991-03-18 2007-03-20 Centocor, Inc. Methods of treating psoriasis with anti-TNF antibodies
US6136310A (en) * 1991-07-25 2000-10-24 Idec Pharmaceuticals Corporation Recombinant anti-CD4 antibodies for human therapy
JPH05244982A (ja) 1991-12-06 1993-09-24 Sumitomo Chem Co Ltd 擬人化b−b10
US5777085A (en) 1991-12-20 1998-07-07 Protein Design Labs, Inc. Humanized antibodies reactive with GPIIB/IIIA
IL101769A (en) 1992-05-03 2007-02-11 Yeda Res & Dev Modulation of TNF receptor action
US6270766B1 (en) 1992-10-08 2001-08-07 The Kennedy Institute Of Rheumatology Anti-TNF antibodies and methotrexate in the treatment of arthritis and crohn's disease
AU5152293A (en) 1992-10-08 1994-05-09 Kennedy Institute Of Rheumatology, The Treatment of autoimmune and inflammatory disorders
PT614984E (pt) 1993-03-05 2001-12-28 Bayer Ag Anticorpos humanos anti-tnf
EP0631783A1 (fr) 1993-06-03 1995-01-04 Mitsubishi Chemical Corporation Combinaisons antivirales de 2',3'-didéoxy-ribonucléosides avec des dérivés 5-substitués du 6-benzyl-1-éthoxyméthyl-uracile
JPH09510952A (ja) 1993-10-06 1997-11-04 ザ ケネディー インスティチュート オブ リューマトロジー 自己免疫疾患および炎症性疾患の治療
US20020068057A1 (en) 1994-03-10 2002-06-06 Marc Feldmann Treatment of autoimmune and inflammatory disorders
PT840618E (pt) 1995-05-18 2003-08-29 Ortho Mcneil Pharm Inc Inducao de tolerancia imunologica atraves de anticorpos anti-cd4 nao-depletores
DK0929578T3 (da) 1996-02-09 2003-08-25 Abbott Lab Bermuda Ltd Humane antistoffer, der binder human TNFalfa
US20010056066A1 (en) 1996-07-26 2001-12-27 Smithkline Beecham Corporation Method of treating immune cell mediated systemic diseases
WO1998014211A1 (fr) 1996-10-01 1998-04-09 Celltech Therapeutics Limited Produits pharmaceutiques contenant un inhibiteur de la kinase agissant sur la tyrosine de proteines et des anticorps anti-cd4
DE19722888A1 (de) 1997-05-28 1998-12-03 Thomas Prof Dr Huenig Human-CD28 spezifische monoklonale Antikörper zur antigenunspezifischen Aktivierung von T-Lymphozyten
WO2000058362A1 (fr) * 1999-03-26 2000-10-05 Human Genome Sciences, Inc. Nouvelles proteines capables de liaison avec la neutrokine alpha et procedes bases sur lesdites proteines
US7074403B1 (en) 1999-06-09 2006-07-11 Immunomedics, Inc. Immunotherapy of autoimmune disorders using antibodies which target B-cells
EP1289554A4 (fr) 2000-06-02 2004-05-26 Univ Minnesota Procede immunotherapeutique permettant de prevenir le rejet de cellules insulaires
US20020058029A1 (en) 2000-09-18 2002-05-16 Nabil Hanna Combination therapy for treatment of autoimmune diseases using B cell depleting/immunoregulatory antibody combination
DE10050935A1 (de) 2000-10-11 2002-05-02 Tegenero Gmbh Verwendung CD28 spezifischer monoklonaler Antikörper zur Stimulation von Blutzellen, welche kein CD28 tragen
US6726923B2 (en) 2001-01-16 2004-04-27 Vascular Therapies, Llc Apparatus and methods for preventing or treating failure of hemodialysis vascular access and other vascular grafts
JP4295513B2 (ja) 2001-03-07 2009-07-15 チルドレンズ メディカル センター コーポレーション ミニ細胞ディスプレイを用いるペプチドディスプレイライブラリーをスクリーニングする方法
EP1241249A1 (fr) 2001-03-12 2002-09-18 Gerold Schuler Lymphocytes T CD4+CD25+ régulateurs provenant du sang humain
JP4448906B2 (ja) 2001-06-14 2010-04-14 イシス イノベーション リミテッド Cd4特異的抗体trx1およびその使用
US7541443B2 (en) * 2001-06-14 2009-06-02 Tolerrx, Inc. Anti-CD4 antibodies
GB2376466A (en) 2001-06-14 2002-12-18 Mark Frewin TRX1 antibody
AU2002357427A1 (en) 2001-12-04 2003-06-17 Tegenero Ag Peptide or protein containing a c'-d loop of the cd28 receptor family
DE10212108A1 (de) 2002-03-13 2003-10-02 Tegenero Ag Verwendung einer an CD28 bindenden Wirksubstanz zur Herstellung einer pharmazeutischen Zusammensetzung
DE10230223A1 (de) 2002-07-04 2004-01-22 Tegenero Ag Mikropartikel mit CD28-spezifischen monoklonalen Antikörpern
AU2003291625B2 (en) 2002-09-16 2009-10-08 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
US7388079B2 (en) 2002-11-27 2008-06-17 The Regents Of The University Of California Delivery of pharmaceutical agents via the human insulin receptor
US7501494B2 (en) 2003-01-15 2009-03-10 United Biomedical, Inc. Designed deimmunized monoclonal antibodies for protection against HIV exposure and treatment of HIV infection
EP1460088A1 (fr) * 2003-03-21 2004-09-22 Biotest AG Anticorps humanisé contre CD4 avec des caractéristiques immunosuppressives
ES2322267T3 (es) 2003-04-09 2009-06-18 Genentech, Inc. Terapia de una enfermedad autoinmunologica en un paciente que presenta una respuesta inadecuada a un inhibidor de tnf-alfa.
GB0314461D0 (en) * 2003-06-20 2003-07-23 Isis Innovation Suppression of transplant rejection
AU2003264100A1 (en) 2003-08-25 2005-03-10 Pieris Proteolab Ag Muteins of a bilin-binding protein with affinity for a given target
EP1600164A3 (fr) 2003-09-22 2006-05-17 TeGenero AG Utilisation d'une substance liant le CD28 dans la production d'une composition pharmaceutique à effet dose-dépendant
DE10352900A1 (de) 2003-11-11 2005-06-16 Tegenero Ag Verwendung einer an CD28 bindenden Wirksubstanz zur Herstellung einer pharmazeutischen Zusammensetzung zur Behandlung von B-CLL
EP1758936A2 (fr) 2004-06-22 2007-03-07 Tolerrx Inc. Dosage optimise d'anticorps anti-cd4 inducteurs de tolerance chez des primates
US20070270431A1 (en) 2004-09-29 2007-11-22 Kowa Co., Ltd. Preventive and/or Therapeutic Medicine for Rheumatoid Arthritis
PT2171060E (pt) 2004-11-11 2013-11-18 Theramab Llc Anticorpo anti-cd28 superagonista
WO2006055077A2 (fr) 2004-11-12 2006-05-26 The University Of North Carolina At Charlotte Modulation de reponses de lymphocytes t cd4+ par une proteine salivaire de tique, salp15 et polypeptides derives de celle-ci
DE102004063494A1 (de) 2004-12-23 2006-07-13 Tegenero Ag Antikörper
JP4730733B2 (ja) 2005-05-02 2011-07-20 国立大学法人京都大学 4型葉酸受容体の発現を指標とした制御性t細胞の検出方法、及び免疫賦活剤
US8663634B2 (en) * 2005-07-11 2014-03-04 Macrogenics, Inc. Methods for the treatment of autoimmune disorders using immunosuppressive monoclonal antibodies with reduced toxicity
JP5230420B2 (ja) 2005-08-18 2013-07-10 ゲンマブ エー/エス Cd4結合ペプチドおよび放射線による治療
BRPI0615397B1 (pt) 2005-08-26 2023-10-03 Roche Glycart Ag Anticorpo anti-cd20, composição farmacêutica que o contém e uso do mesmo
SI1973950T1 (sl) 2006-01-05 2015-01-30 Genentech, Inc. Anti-EphB4 protitelesa in postopki njihove uporabe
MX2008011785A (es) 2006-03-16 2008-09-25 Genentech Inc Metodos de tratamiento de lupus utilizando anticuerpos cd4.
EP2010569A4 (fr) 2006-03-20 2009-09-09 Xoma Technology Ltd Anticorps humains spécifiques à des matières et des procédés à base de gastrines
US20090208471A1 (en) 2006-04-07 2009-08-20 Yun Theodore J Isolation and Use of Human Regulatory T Cells
WO2007135684A2 (fr) 2006-05-22 2007-11-29 Hadasit Medical Research Services & Development Limited Procédé de traitement d'auto-immunité anti-cd4
ES2593789T3 (es) 2007-02-01 2016-12-13 Universitätsmedizin Der Johannes Gutenberg-Universität Mainz Activación específica de una célula T reguladora y su uso para el tratamiento del asma, enfermedades alérgicas, enfermedades autoinmunes, rechazo de injertos y para la inducción de tolerancia
EP2599791A1 (fr) 2007-04-27 2013-06-05 Genentech, Inc. Anticorps anti-CD4 puissants, stables et non immunosuppresseurs
ES2610327T3 (es) 2008-03-13 2017-04-27 Biotest Ag Régimen de dosificación de anticuerpos anti-CD4 para el tratamiento de enfermedades autoinmunitarias
BRPI0909180A2 (pt) 2008-03-13 2016-08-09 Biotest Ag composição farmacêutica, e, método de tratamento de uma doença autoimune
EP2101173A1 (fr) 2008-03-14 2009-09-16 Vivalis Procédé in vitro pour déterminer si un candidat destiné à un médicament pour une protéine cible est actif contre une variante de cette protéine
WO2010022341A1 (fr) 2008-08-21 2010-02-25 The United State Of America, As Represented By The Secretary, Department Of Health And Human Services Procédés pour enrichir et utiliser des cellules t régulatrices
ES2335381B1 (es) 2008-09-24 2011-02-28 Fundacio Institut De Recerca De L'hospital Universitari Vall D'hebron Metodo in vitro y kit para el pronostico o prediccion de la respuestapor parte de pacientes con artritis reumatoide al tratamiento con agentes bloqueantes del factor tnfalfa.
MX2011003335A (es) 2008-09-29 2011-04-27 Biotest Ag Composicion para tratar enfermedad.

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ANONYMOUS: "Tregalizumab (BT-061) Shows Efficacy in Chronic Plaque Psoriasis", 24 November 2011 (2011-11-24), pages 1 - 2, XP055037178, Retrieved from the Internet <URL:http://www.evaluatepharma.com/Universal/View.aspx?type=Story&id=266131> [retrieved on 20120904] *

Also Published As

Publication number Publication date
US20110059084A1 (en) 2011-03-10
KR20100135257A (ko) 2010-12-24
BRPI0909048A2 (pt) 2015-11-24
CA2718184A1 (fr) 2009-10-08
CR11724A (es) 2011-06-07
ZA201007215B (en) 2016-08-31
AU2009231325B2 (en) 2014-09-18
JP5597553B2 (ja) 2014-10-01
IL207891A0 (en) 2010-12-30
EP2265643A1 (fr) 2010-12-29
US9334325B2 (en) 2016-05-10
JP2011516416A (ja) 2011-05-26
MX2010010028A (es) 2011-08-17
CO6311007A2 (es) 2011-08-22
RU2010141826A (ru) 2012-04-20
SG190598A1 (en) 2013-06-28
CN102027017A (zh) 2011-04-20
RU2539110C2 (ru) 2015-01-10
WO2009121690A1 (fr) 2009-10-08
AU2009231325A1 (en) 2009-10-08

Similar Documents

Publication Publication Date Title
EP2265643B1 (fr) Schema posologique pour traiter le psoriasis et l&#39;arhtrite rhumatoide
EP2262838B1 (fr) Agent pour traiter une maladie
EP2260057B1 (fr) Régime posologique d&#39;un anticorps anti-CD4 pour le traitement de maladies auto-immunes
EP2332991B1 (fr) Anticorps anti-CD4 humanisé doté de propriétés immunosuppressives
MX2011003335A (es) Composicion para tratar enfermedad.

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20101013

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA RS

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1151047

Country of ref document: HK

17Q First examination report despatched

Effective date: 20120918

RIN1 Information on inventor provided before grant (corrected)

Inventor name: GERMER, MATTHIAS

Inventor name: KRAUS, ELMAR

Inventor name: WARTENBERG-DEMAND, ANDREA

Inventor name: KAISER, SIBYLLE

Inventor name: DAELKEN, BENJAMIN

Inventor name: AIGNER, SILKE

Inventor name: UHEREK, CHRISTOPH

Inventor name: SCHULZ, GREGOR

Inventor name: WOLF, DANIELE

Inventor name: BRUECHER, CHRISTOPH

Inventor name: LINDNER, JUERGEN

Inventor name: OSTERROTH, FRANK

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20160127

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAL Information related to payment of fee for publishing/printing deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR3

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20160706

INTG Intention to grant announced

Effective date: 20160711

INTG Intention to grant announced

Effective date: 20160804

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 838255

Country of ref document: AT

Kind code of ref document: T

Effective date: 20161115

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602009041827

Country of ref document: DE

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 9

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20161019

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 838255

Country of ref document: AT

Kind code of ref document: T

Effective date: 20161019

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170120

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170119

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170220

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170219

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602009041827

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170119

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

26N No opposition filed

Effective date: 20170720

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1151047

Country of ref document: HK

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170310

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 10

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170310

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170331

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170331

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170310

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602009041827

Country of ref document: DE

Representative=s name: PAGE, WHITE & FARRER GERMANY LLP, DE

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20190306

Year of fee payment: 11

Ref country code: DE

Payment date: 20190226

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20190213

Year of fee payment: 11

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20090310

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20161019

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20161019

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602009041827

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201001

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200331

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20200310

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200310